Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Ann Neurol ; 77(5): 784-803, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25628066

RESUMO

OBJECTIVE: Recent evidence suggests that ischemic stroke is a thromboinflammatory disease. Plasma kallikrein (PK) cleaves high-molecular-weight kininogen to release bradykinin (BK) and is a key constituent of the proinflammatory contact-kinin system. In addition, PK can activate coagulation factor XII, the origin of the intrinsic coagulation cascade. Thus, PK triggers 2 important pathological pathways of stroke formation, thrombosis and inflammation. METHODS: We investigated the consequences of PK inhibition in transient and permanent models of ischemic stroke. RESULTS: PK-deficient mice of either sex challenged with transient middle cerebral artery occlusion developed significantly smaller brain infarctions and less severe neurological deficits compared with controls without an increase in infarct-associated hemorrhage. This protective effect was preserved at later stages of infarctions as well as after permanent stroke. Reduced intracerebral thrombosis and improved cerebral blood flow could be identified as underlying mechanisms. Moreover, blood-brain barrier function was maintained in mice lacking PK, and the local inflammatory response was reduced. PK-deficient mice reconstituted with PK or BK again developed brain infarctions similar to wild-type mice. Important from a translational perspective, inhibition of PK in wild-type mice using a PK-specific antibody was likewise effective even when performed in a therapeutic setting up to 3 hours poststroke. INTERPRETATION: PK drives thrombus formation and inflammation via activation of the intrinsic coagulation cascade and the release of BK but appears to be dispensable for hemostasis. Hence, PK inhibition may offer a safe strategy to combat thromboembolic disorders including ischemic stroke.


Assuntos
Calicreína Plasmática/metabolismo , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/prevenção & controle , Trombose/sangue , Trombose/prevenção & controle , Animais , Infarto Encefálico/sangue , Infarto Encefálico/genética , Infarto Encefálico/prevenção & controle , Feminino , Inflamação/sangue , Inflamação/genética , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Calicreína Plasmática/antagonistas & inibidores , Calicreína Plasmática/genética , Acidente Vascular Cerebral/genética , Trombose/genética
2.
Pflugers Arch ; 467(5): 973-87, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25315980

RESUMO

Potassium channels can fulfill both beneficial and detrimental roles in neuronal damage during ischemic stroke. Earlier studies have characterized a neuroprotective role of the two-pore domain potassium channels KCNK2 (TREK1) and KCNK3 (TASK1). Protective neuronal hyperpolarization and prevention of intracellular Ca(2+) overload and glutamate excitotoxicity were suggested to be the underlying mechanisms. We here identify an unexpected role for the related KCNK5 channel in a mouse model of transient middle cerebral artery occlusion (tMCAO). KCNK5 is strongly upregulated on neurons upon cerebral ischemia, where it is most likely involved in the induction of neuronal apoptosis. Hypoxic conditions elevated neuronal expression levels of KCNK5 in acute brain slices and primary isolated neuronal cell cultures. In agreement, KCNK5 knockout mice had significantly reduced infarct volumes and improved neurologic function 24 h after 60 min of tMCAO and this protective effect was preserved at later stages of infarct development. KCNK5 deficiency resulted in a significantly reduced number of apoptotic neurons, a downregulation of pro-apoptotic and upregulation of anti-apoptotic factors. Results of adoptive transfer experiments of wild-type and Kcnk5 (-/-) immune cells into Rag1 (-/-) mice prior to tMCAO exclude a major role of KCNK5 in poststroke inflammatory reactions. In summary, KCNK5 expression is induced on neurons under ischemic conditions where it most likely exerts pro-apoptotic effects. Hence, pharmacological blockade of KCNK5 might have therapeutic potential in preventing ischemic neurodegeneration.


Assuntos
Isquemia Encefálica/metabolismo , Neurônios/metabolismo , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Humanos , Infarto da Artéria Cerebral Média/fisiopatologia , Acidente Vascular Cerebral/fisiopatologia
3.
Blood ; 121(4): 679-91, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23160472

RESUMO

We have recently identified T cells as important mediators of ischemic brain damage, but the contribution of the different T-cell subsets is unclear. Forkhead box P3 (FoxP3)-positive regulatory T cells (Tregs) are generally regarded as prototypic anti-inflammatory cells that maintain immune tolerance and counteract tissue damage in a variety of immune-mediated disorders. In the present study, we examined the role of Tregs after experimental brain ischemia/reperfusion injury. Selective depletion of Tregs in the DEREG mouse model dramatically reduced infarct size and improved neurologic function 24 hours after stroke and this protective effect was preserved at later stages of infarct development. The specificity of this detrimental Treg effect was confirmed by adoptive transfer experiments in wild-type mice and in Rag1(-/-) mice lacking lymphocytes. Mechanistically, Tregs induced microvascular dysfunction in vivo by increased interaction with the ischemic brain endothelium via the LFA-1/ICAM-1 pathway and platelets and these findings were confirmed in vitro. Ablation of Tregs reduced microvascular thrombus formation and improved cerebral reperfusion on stroke, as revealed by ultra-high-field magnetic resonance imaging at 17.6 Tesla. In contrast, established immunoregulatory characteristics of Tregs had no functional relevance. We define herein a novel and unexpected role of Tregs in a primary nonimmunologic disease state.


Assuntos
Isquemia Encefálica/imunologia , Microvasos/fisiopatologia , Acidente Vascular Cerebral/metabolismo , Linfócitos T Reguladores/metabolismo , Transferência Adotiva , Animais , Plaquetas/imunologia , Plaquetas/metabolismo , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/genética , Isquemia Encefálica/terapia , Comunicação Celular , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Depleção Linfocítica , Masculino , Camundongos , Camundongos Knockout , Microvasos/patologia , Acidente Vascular Cerebral/imunologia , Acidente Vascular Cerebral/terapia , Linfócitos T Reguladores/imunologia
4.
Blood ; 120(19): 4082-92, 2012 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-22936662

RESUMO

Thrombosis and inflammation are hallmarks of ischemic stroke still unamenable to therapeutic interventions. High-molecular-weight kininogen (KNG) is a central constituent of the contact-kinin system which represents an interface between thrombotic and inflammatory circuits and is critically involved in stroke development. Kng(-/-) mice are protected from thrombosis after artificial vessel wall injury and lack the proinflammatory mediator bradykinin. We investigated the consequences of KNG deficiency in models of ischemic stroke. Kng(-/-) mice of either sex subjected to transient middle cerebral artery occlusion developed dramatically smaller brain infarctions and less severe neurologic deficits without an increase in infarct-associated hemorrhage. This protective effect was preserved at later stages of infarction as well as in elderly mice. Targeting KNG reduced thrombus formation in ischemic vessels and improved cerebral blood flow, and reconstitution of KNG-deficient mice with human KNG or bradykinin restored clot deposition and infarct susceptibility. Moreover, mice deficient in KNG showed less severe blood-brain barrier damage and edema formation, and the local inflammatory response was reduced compared with controls. Because KNG appears to be instrumental in pathologic thrombus formation and inflammation but dispensable for hemostasis, KNG inhibition may offer a selective and safe strategy for combating stroke and other thromboembolic diseases.


Assuntos
Barreira Hematoencefálica/fisiopatologia , Isquemia Encefálica/prevenção & controle , Cininogênios/deficiência , Trombose/fisiopatologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Edema Encefálico/genética , Edema Encefálico/prevenção & controle , Isquemia Encefálica/genética , Isquemia Encefálica/mortalidade , Modelos Animais de Doenças , Feminino , Inflamação/genética , Inflamação/patologia , Hemorragias Intracranianas/diagnóstico , Cininogênios/genética , Cininogênios/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Knockout , Fluxo Sanguíneo Regional , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/mortalidade , Acidente Vascular Cerebral/prevenção & controle , Trombose/genética
5.
Stroke ; 44(11): 3202-10, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24029635

RESUMO

BACKGROUND AND PURPOSE: Lymphocytes are important players in the pathophysiology of acute ischemic stroke. The interaction of lymphocytes with endothelial cells and platelets, termed thrombo-inflammation, fosters microvascular dysfunction and secondary infarct growth. FTY720, a sphingosine-1-phosphate receptor modulator, blocks the egress of lymphocytes from lymphoid organs and has been shown to reduce ischemic neurodegeneration; however, the underlying mechanisms are unclear. We investigated the mode of FTY720 action in models of cerebral ischemia. METHODS: Transient middle cerebral artery occlusion (tMCAO) was induced in wild-type and lymphocyte-deficient Rag1(-/-) mice treated with FTY720 (1 mg/kg) or vehicle immediately before reperfusion. Stroke outcome was assessed 24 hours later. Immune cells in the blood and brain were counted by flow cytometry. The integrity of the blood-brain barrier was analyzed using Evans Blue dye. Thrombus formation was determined by immunohistochemistry and Western blot, and was correlated with cerebral perfusion. RESULTS: FTY720 significantly reduced stroke size and improved functional outcome in wild-type mice on day 1 and day 3 after transient middle cerebral artery occlusion. This protective effect was lost in lymphocyte-deficient Rag1(-/-) mice and in cultured neurons subjected to hypoxia. Less lymphocytes were present in the cerebral vasculature of FTY720-treated wild-type mice, which in turn reduced thrombosis and increased cerebral perfusion. In contrast, FTY720 was unable to prevent blood-brain barrier breakdown and transendothelial immune cell trafficking after transient middle cerebral artery occlusion. CONCLUSIONS: Induction of lymphocytopenia and concomitant reduction of microvascular thrombosis are key modes of FTY720 action in stroke. In contrast, our findings in Rag1(-/-) mice and cultured neurons argue against direct neuroprotective effects of FTY720.


Assuntos
Isquemia Encefálica/terapia , Imunossupressores/farmacologia , Inflamação/terapia , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Acidente Vascular Cerebral/terapia , Trombose/terapia , Animais , Inibidores Enzimáticos/farmacologia , Cloridrato de Fingolimode , Proteínas de Homeodomínio/genética , Hipóxia , Linfócitos/efeitos dos fármacos , Linfopenia/patologia , Masculino , Camundongos , Camundongos Transgênicos , Artéria Cerebral Média/patologia , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Esfingosina/farmacologia
6.
Nat Genet ; 31(3): 311-5, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12068299

RESUMO

A substantial percentage of human pregnancies are lost as spontaneous abortions after implantation. This is often caused by an inadequately developed placenta. Proper development of the placental vascular system is essential to nutrient and gas exchange between mother and developing embryo. Here we show that alpha(2)-adrenoceptors, which are activated by adrenaline and noradrenaline, are important regulators of placental structure and function. Mice with deletions in the genes encoding alpha(2A)-, alpha(2B)- and alpha(2C)-adrenoceptors died between embryonic days 9.5 and 11.5 from a severe defect in yolk-sac and placenta development. In wildtype placentae, alpha(2)-adrenoceptors are abundantly expressed in giant cells, which secrete angiogenic factors to initiate development of the placental vascular labyrinth. In placentae deficient in alpha(2A)-, alpha(2B)- and alpha(2C)-adrenoceptors, the density of fetal blood vessels in the labyrinth was markedly lower than normal, leading to death of the embryos as a result of reduced oxygen and nutrient supply. Basal phosphorylation of the extracellular signal regulated kinases ERK1 and ERK2 was also lower than normal, suggesting that activation of the mitogen-activated protein kinase (MAP kinase) pathway by alpha(2)-adrenoceptors is required for placenta and yolk-sac vascular development. Thus, alpha(2)-adrenoceptors are essential at the placental interface between mother and embryo to establish the circulatory system of the placenta and thus maintain pregnancy.


Assuntos
Placenta/metabolismo , Receptores Adrenérgicos alfa 2/fisiologia , Animais , AMP Cíclico/análise , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário e Fetal/genética , Feminino , Morte Fetal/genética , Coração Fetal/embriologia , Deleção de Genes , Camundongos , Camundongos Mutantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Placenta/irrigação sanguínea , Gravidez , Receptores Adrenérgicos alfa 2/deficiência
7.
Stroke ; 43(9): 2457-67, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22744646

RESUMO

BACKGROUND AND PURPOSE: Inflammation and thrombosis are pathophysiological hallmarks of ischemic stroke still unamenable to therapeutic interventions. The contact-kinin system represents an interface between inflammatory and thrombotic circuits and is involved in stroke development. C1-inhibitor counteracts activation of the contact-kinin system at multiple levels. We investigated the therapeutic potential of C1-inhibitor in models of ischemic stroke. METHODS: Male and female C57Bl/6 mice and rats of different ages were subjected to middle cerebral artery occlusion and treated with C1-inhibitor after 1 hour or 6 hours. Infarct volumes and functional outcomes were assessed between day 1 and day 7, and findings were validated by magnetic resonance imaging. Blood-brain barrier damage, thrombus formation, and the local inflammatory response were determined poststroke. RESULTS: Treatment with 15.0 U C1-inhibitor, but not 7.5 U, 1 hour after stroke reduced infarct volumes by ≈60% and improved clinical scores in mice of either sex on day 1. This protective effect was preserved at later stages of infarction as well as in elderly mice and in another species, ie, rats. Delayed C1-inhibitor treatment still improved clinical outcome. Blood-brain barrier damage, edema formation, and inflammation were significantly lower compared with controls. Moreover, C1-inhibitor showed strong antithrombotic effects. CONCLUSIONS: C1-inhibitor is a multifaceted antiinflammatory and antithrombotic compound that protects from ischemic neurodegeneration in clinically meaningful settings.


Assuntos
Anti-Inflamatórios , Isquemia Encefálica/prevenção & controle , Proteína Inibidora do Complemento C1/uso terapêutico , Fibrinolíticos , Traumatismo por Reperfusão/prevenção & controle , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Western Blotting , Edema Encefálico/tratamento farmacológico , Edema Encefálico/patologia , Isquemia Encefálica/patologia , Feminino , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Trombose Intracraniana/tratamento farmacológico , Trombose Intracraniana/patologia , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Exame Neurológico , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Traumatismo por Reperfusão/patologia , Caracteres Sexuais , Resultado do Tratamento
8.
Saudi J Anaesth ; 15(2): 127-130, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34188629

RESUMO

BACKGROUND: Adequate preoperative information can lessen patient anxiety. Delivering sufficient information during a personal interview, however, is time consuming, and therefore a relevant economical aspect. We investigated whether video information given to the patient before the pre-anesthetic interview has an influence on the patient's anxiety and the duration of the interview. METHOD: We randomized 302 patients undergoing different types of anesthesia. In all, 151 patients watched a short video with general information about the anticipated anesthesia procedure. Afterward, all patients had a standard pre-anesthetic interview. Patients' anxiety and satisfaction with pre-anesthesia care were assessed after the interview using a visual analogue scale. The duration of the interview was documented. Student t-test and P < 0.05 for differences between the groups. RESULTS: There was no difference in gender, age, ASA physical status, previous anesthesia experience, and the planned anesthesia procedure between the two groups. No difference in anxiety and satisfaction with pre-anesthesia care was observed. The duration of the pre-anesthetic interview was also not different between the groups. DISCUSSION: Preoperative multimedia information did not reduce anxiety or increase the patient satisfaction undergoing anesthesia. The video containing general information did not save time in the pre-anesthetic interview.

9.
Mol Pharmacol ; 75(5): 1160-70, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19251826

RESUMO

Alpha(2)-adrenoceptors mediate diverse functions of the sympathetic system and are targets for the treatment of cardiovascular disease, depression, pain, glaucoma, and sympathetic activation during opioid withdrawal. To determine whether alpha(2)-adrenoceptors on adrenergic neurons or alpha(2)-adrenoceptors on nonadrenergic neurons mediate the physiological and pharmacological responses of alpha(2)-agonists, we used the dopamine beta-hydroxylase (Dbh) promoter to drive expression of alpha(2A)-adrenoceptors exclusively in noradrenergic and adrenergic cells of transgenic mice. Dbh-alpha(2A) transgenic mice were crossed with double knockout mice lacking both alpha(2A)- and alpha(2C)-receptors to generate lines with selective expression of alpha(2A)-autoreceptors in adrenergic cells. These mice were subjected to a comprehensive phenotype analysis and compared with wild-type mice, which express alpha(2A)- and alpha(2C)-receptors in both adrenergic and nonadrenergic cells, and alpha(2A)/alpha(2C) double-knockout mice, which do not express these receptors in any cell type. We were surprised to find that only a few functions previously ascribed to alpha(2)-adrenoceptors were mediated by receptors on adrenergic neurons, including feedback inhibition of norepinephrine release from sympathetic nerves and spontaneous locomotor activity. Other agonist effects, including analgesia, hypothermia, sedation, and anesthetic-sparing, were mediated by alpha(2)-receptors in nonadrenergic cells. In dopamine beta-hydroxylase knockout mice lacking norepinephrine, the alpha(2)-agonist medetomidine still induced a loss of the righting reflex, confirming that the sedative effect of alpha(2)-adrenoceptor stimulation is not mediated via autoreceptor-mediated inhibition of norepinephrine release. The present study paves the way for a revision of the current view of the alpha(2)-adrenergic receptors, and it provides important new considerations for future drug development.


Assuntos
Receptores Adrenérgicos alfa 2/fisiologia , Agonistas alfa-Adrenérgicos/farmacologia , Analgésicos/farmacologia , Anestésicos/farmacologia , Animais , Temperatura Corporal/efeitos dos fármacos , Hipnóticos e Sedativos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Norepinefrina/metabolismo , Receptores Adrenérgicos alfa 2/genética , Transgenes
10.
Stroke ; 40(1): 285-93, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18988906

RESUMO

BACKGROUND AND PURPOSE: Brain edema is detrimental in ischemic stroke and its treatment options are limited. Kinins are proinflammatory peptides that are released during tissue injury. The effects of kinins are mediated by 2 different receptors (B1 and B2 receptor [B1R and B2R]) and comprise induction of edema formation and release of proinflammatory mediators. METHODS: Focal cerebral ischemia was induced in B1R knockout, B2R knockout, and wild-type mice by transient middle cerebral artery occlusion. Infarct volumes were measured by planimetry. Evan's blue tracer was applied to determine the extent of brain edema. Postischemic inflammation was assessed by real-time reverse-transcriptase polymerase chain reaction and immunohistochemistry. To analyze the effect of a pharmacological kinin receptor blockade, B1R and B2R inhibitors were injected. RESULTS: B1R knockout mice developed significantly smaller brain infarctions and less neurological deficits compared to wild-type controls (16.8+/-4.7 mm(3) vs 50.1+/-9.1 mm(3), respectively; P<0.0001). This was accompanied by a dramatic reduction of brain edema and endothelin-1 expression, as well as less postischemic inflammation. Pharmacological blockade of B1R likewise salvaged ischemic tissue (15.0+/-9.5 mm(3) vs 50.1+/-9.1 mm(3), respectively; P<0.01) in a dose-dependent manner, even when B1R inhibitor was applied 1 hour after transient middle cerebral artery occlusion. In contrast, B2R deficiency did not confer neuroprotection and had no effect on the development of tissue edema. CONCLUSIONS: These data demonstrate that blocking of B1R can diminish brain infarction and edema formation in mice and may open new avenues for acute stroke treatment in humans.


Assuntos
Antagonistas de Receptor B1 da Bradicinina , Bradicinina/análogos & derivados , Edema Encefálico/tratamento farmacológico , Infarto Cerebral/tratamento farmacológico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Animais , Bradicinina/antagonistas & inibidores , Bradicinina/metabolismo , Bradicinina/farmacologia , Bradicinina/uso terapêutico , Antagonistas de Receptor B2 da Bradicinina , Edema Encefálico/metabolismo , Edema Encefálico/fisiopatologia , Artérias Cerebrais/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Artérias Cerebrais/fisiopatologia , Infarto Cerebral/metabolismo , Infarto Cerebral/fisiopatologia , Circulação Cerebrovascular/efeitos dos fármacos , Circulação Cerebrovascular/fisiologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Regulação para Baixo/genética , Encefalite/tratamento farmacológico , Encefalite/fisiopatologia , Encefalite/prevenção & controle , Endotelina-1/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Receptor B1 da Bradicinina/genética , Receptor B1 da Bradicinina/metabolismo , Receptor B2 da Bradicinina/genética , Receptor B2 da Bradicinina/metabolismo
11.
Cardiovasc Res ; 75(4): 728-37, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17597596

RESUMO

OBJECTIVE: Feedback regulation of norepinephrine release from sympathetic nerves is essential to control blood pressure, heart rate and contractility. Recent experiments in gene-targeted mice have suggested that alpha(2C)-adrenoceptors may operate in a similar feedback mechanism to control the release of epinephrine from the adrenal medulla. As heterozygous polymorphisms in the human alpha(2C)-adrenoceptor gene have been associated with cardiovascular disease including hypertension and chronic heart failure, we have sought to characterize the relevance of alpha(2C)-gene copy number for feedback control of epinephrine release in gene-targeted mice. METHODS: Adrenal catecholamine release, basal hemodynamics and susceptibility to develop heart failure after transverse aortic constriction were tested in mice with two copies (+/+), one copy (+/-) or no functional alpha(2C)-adrenoceptor gene (alpha(2C)-/-). RESULTS: Heterozygous alpha(2C)-receptor deletion (alpha(2C)+/-) resulted in a 43% reduction of adrenal alpha(2C) mRNA copy number and in a similar decrease in alpha(2)-receptor-mediated inhibition of catecholamine release from isolated adrenal glands in vitro. Urinary excretion of epinephrine was increased by 74+/-15% in alpha(2C)+/- and by 142+/-23% in alpha(2C)-/- mice as compared with wild-type control mice. Telemetric determination of cardiovascular function revealed significant tachycardia but no hypertension in alpha(2C)-adrenoceptor-deficient mice. alpha(2C)+/- mice were more susceptible to develop cardiac hypertrophy, failure and mortality after left-ventricular pressure overload than alpha(2C)+/+ mice. CONCLUSION: Adrenal alpha(2)-mediated feedback regulation of epinephrine secretion differs fundamentally from sympathetic feedback control. A single adrenoceptor subtype, alpha(2C), operates without a significant receptor reserve to prevent elevation of circulating epinephrine levels. This genetic model may provide an experimental basis to study the pathophysiology of alpha(2C)-adrenoceptor dysfunction in humans.


Assuntos
Medula Suprarrenal/metabolismo , Epinefrina/metabolismo , Insuficiência Cardíaca/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Animais , Aorta , Pressão Sanguínea , Progressão da Doença , Epinefrina/análise , Epinefrina/urina , Retroalimentação Fisiológica , Frequência do Gene , Engenharia Genética , Predisposição Genética para Doença , Insuficiência Cardíaca/genética , Frequência Cardíaca , Heterozigoto , Rim/metabolismo , Masculino , Camundongos , Camundongos Knockout , Polimorfismo Genético , Receptores Adrenérgicos alfa 2/genética , Telemetria , Técnicas de Cultura de Tecidos , Vasoconstrição
12.
Anesth Analg ; 105(6): 1644-9, table of contents, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18042862

RESUMO

BACKGROUND: The IV anesthetic, etomidate, has structural and clinical similarities to specific alpha2-adrenoceptor agonists such as dexmedetomidine. We investigated whether the sedative effects of etomidate may be mediated by alpha2-adrenoceptors. METHODS: The anesthetic potency of etomidate (1-20 microM) was determined in Xenopus laevis tadpoles in the absence and presence of the specific alpha2-adrenoceptor antagonist atipamezole (10 microM). Anesthesia was defined as loss of righting reflex. Nonlinear logistic regression curves were fitted to the data and half-maximal effective concentrations and the slopes of the curves were calculated. Additionally, sedative/ hypnotic effects of etomidate (8 mg/kg IP) were studied by rotarod test in wild-type (WT) mice and mice carrying targeted deletions of the alpha2A-adrenoceptor gene (alpha2A-KO). Data are presented as mean +/- sem. RESULTS: The fraction of anesthetized tadpoles increased with increasing concentrations of etomidate. Atipamezole significantly increased the half-maximal effective concentration of etomidate (4.5 +/- 0.2 microM; slope: 2.6 +/- 0.3) to 8.4 +/- 0.4 microM (slope: 2.3 +/- 0.3). Etomidate resulted in time-dependent sedative effects in all mice, as assessed by rotarod performance. In WT mice, the sedative effects of etomidate were not decreased by atipamezole (2 mg/kg). Consistently, etomidate-induced sedation was not reduced in alpha2A-KO animals compared with WT mice. CONCLUSIONS: The sedative effects of etomidate exhibit a species-specific interaction with alpha2-adrenoceptors. Although the decrease in potency of etomidate by atipamezole may be caused by an interaction with alpha2-adrenoceptors in X. laevis tadpoles, results in mice indicate that the hypnotic effect of etomidate does not require alpha2-adrenoceptors.


Assuntos
Anestesia , Etomidato/metabolismo , Receptores Adrenérgicos alfa 2/genética , Receptores Adrenérgicos alfa 2/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2 , Anestesia/métodos , Animais , Relação Dose-Resposta a Droga , Etomidato/farmacologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Especificidade da Espécie , Xenopus laevis
13.
Anesth Analg ; 102(2): 456-61, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16428542

RESUMO

We performed the current study in mice lacking individual alpha2-adrenoceptor subtypes to elucidate the contribution of alpha(2)-adrenoceptor subtypes to the neuroprotective properties of dexmedetomidine in a model of perinatal excitotoxic brain injury. On postnatal Day 5, wild-type mice and mice lacking alpha2A-adrenoceptor (alpha2A-KO) or alpha2C-adrenoceptor subtypes (alpha2C-KO) were randomly assigned to receive dexmedetomidine (3 microg/kg) or phosphate-buffered saline intraperitoneally. Thirty minutes after the intraperitoneal injection, the glutamatergic agonist ibotenate (10 microg) was intracerebrally injected, producing transcortical necrosis and white matter lesions that mimic perinatal human hypoxic-like lesions. Quantification of the lesions was performed on postnatal Day 10 by histopathologic examination. Dexmedetomidine reduced mean lesion size in the cortex of wild-type mice and alpha2C-KO mice by 44% and 49%, respectively. Ibotenate-induced white matter lesions were reduced by 71% (wild-type mice) and 75% (alpha2C-KO mice) after pretreatment with dexmedetomidine. In contrast, in alpha2A-KO mice, dexmedetomidine did not protect against the cortical excitotoxic insult, and white matter lesions were even more pronounced (82% increase of mean lesion size). Dexmedetomidine provides potent neuroprotection in a model of perinatal excitotoxic brain damage. This effect was completely abolished in alpha2A-KO mice, suggesting that the neuroprotective effect is mediated via the alpha2A-adrenoceptor subtype.


Assuntos
Agonistas alfa-Adrenérgicos/farmacologia , Encéfalo/patologia , Dexmedetomidina/farmacologia , Agonistas de Aminoácidos Excitatórios/toxicidade , Ácido Ibotênico/toxicidade , Receptores Adrenérgicos alfa 2/metabolismo , Animais , Animais Recém-Nascidos , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Relação Dose-Resposta a Droga , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fármacos Neuroprotetores/farmacologia
14.
Circulation ; 106(19): 2491-6, 2002 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-12417548

RESUMO

BACKGROUND: Elevated plasma norepinephrine levels are associated with increased mortality in patients and in animal models with chronic heart failure. To test which alpha2-adrenoceptor subtypes operate as presynaptic inhibitory receptors to control norepinephrine release in heart failure, we investigated the response of gene-targeted mice lacking alpha2-adrenoceptor subtypes (alpha2-KO) to chronic left ventricular pressure overload. In addition, we determined the functional consequences of genetic variants of alpha2-adrenoceptors in human patients with chronic heart failure. METHODS AND RESULTS: Cardiac pressure overload was induced by transverse aortic constriction. Three months after aortic banding, survival was dramatically reduced in alpha2A-KO (52%) and alpha2C-KO (47%) mice compared with wild-type and alpha2B-deficient (86%) animals. Excess mortality in alpha2A- and alpha2C-KO strains was attributable to heart failure with enhanced left ventricular hypertrophy and fibrosis and elevated circulating catecholamines. The clinical importance of this finding is emphasized by the fact that heart failure patients with a dysfunctional variant of the alpha2C-adrenoceptor had a worse clinical status and decreased cardiac function as determined by invasive catheterization and by echocardiography. CONCLUSIONS: Our results indicate an essential function of alpha2A- and alpha2C-adrenoceptors in the prevention of heart failure progression in mice and human patients. Identification of heart failure patients with genetic alpha2-adrenoceptor variants as well as new alpha2-receptor subtype-selective drugs may represent novel therapeutic strategies in chronic heart failure and other diseases with enhanced sympathetic activation.


Assuntos
Catecolaminas/metabolismo , Retroalimentação Fisiológica , Insuficiência Cardíaca/prevenção & controle , Insuficiência Cardíaca/fisiopatologia , Receptores Adrenérgicos alfa 2/metabolismo , Animais , Estenose da Valva Aórtica/complicações , Estenose da Valva Aórtica/fisiopatologia , Cardiomegalia/etiologia , Cardiomegalia/patologia , Catecolaminas/sangue , Modelos Animais de Doenças , Progressão da Doença , Retroalimentação Fisiológica/fisiologia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/patologia , Hemodinâmica , Humanos , Camundongos , Camundongos Knockout , Norepinefrina/metabolismo , Receptores Adrenérgicos alfa 2/deficiência , Receptores Adrenérgicos alfa 2/genética , Deleção de Sequência , Taxa de Sobrevida
15.
Circulation ; 110(20): 3245-51, 2004 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-15520307

RESUMO

BACKGROUND: Pituitary adenylate cyclase-activating polypeptide (PACAP), acting via 3 different G protein-coupled receptors, has been implicated in the regulation of several homeostatic systems in the body, including cardiopulmonary control. To define the physiologic role of the PACAP-preferring type I receptor, PAC1, in cardiopulmonary function, we developed a mutant mouse strain lacking functional PAC1 receptors. METHODS AND RESULTS: When PAC1-deficient mice were crossed onto a C57BL/6 background, almost all mutants died during the second postnatal week. Whereas mutant mice were indistinguishable from their wild-type littermates at birth, they showed progressive weakness and died from rapidly developing heart failure. Right ventricles of PAC1 mutants were massively dilated and showed cardiac myocyte hypertrophy, whereas left ventricular structure was unaltered. On direct cardiac catheterization, right ventricular pressure was elevated by 45% in PAC1-deficient mice, indicating increased pulmonary artery pressure, as no malformations were detected in the valves or outflow tract of the right ventricle. Consistent with elevated pulmonary pressure, lung capillary density was decreased by 30% and small pulmonary arteries of mutant mice had significant vascular smooth muscle cell hypertrophy compared with wild-type mice. CONCLUSIONS: Whereas PACAP induces vasodilation in isolated pulmonary vessels in wild-type mice, the absence of its specific receptor PAC1 causes pulmonary hypertension and right heart failure after birth. These in vivo findings demonstrate the crucial importance of PAC1-mediated signaling for the maintenance of normal pulmonary vascular tone during early postnatal life.


Assuntos
Insuficiência Cardíaca/genética , Hipertensão Pulmonar/genética , Hipertrofia Ventricular Direita/genética , Fatores de Crescimento Neural/fisiologia , Neuropeptídeos/fisiologia , Neurotransmissores/fisiologia , Receptores de Superfície Celular/fisiologia , Resistência Vascular/fisiologia , Animais , Capilares/patologia , Tamanho Celular , Corticosterona/sangue , Cruzamentos Genéticos , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/metabolismo , Hipertensão Pulmonar/metabolismo , Hipertrofia , Hipertrofia Ventricular Direita/complicações , Hipertrofia Ventricular Direita/diagnóstico por imagem , Hipertrofia Ventricular Direita/metabolismo , Pulmão/irrigação sanguínea , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos Cardíacos/patologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Pressão , Artéria Pulmonar/crescimento & desenvolvimento , Artéria Pulmonar/patologia , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Receptores de Polipeptídeo Hipofisário Ativador de Adenilato Ciclase , Transdução de Sinais , Ultrassonografia , Vasodilatação/genética , Vasodilatação/fisiologia
16.
FASEB J ; 17(2): 283-5, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12490546

RESUMO

UNLABELLED: The role of AT2-receptors has recently been subject of considerable debate. We investigated the influence of AT2-stimulation/inhibition on myocardial endothelial NO-synthase (eNOS, NOS-III) promoter activity and eNOS protein expression. Stimulation of rat cardiomyocytes with angiotensin II (AngII) increased eNOS protein expression 3.3-fold. This was blocked by Cyclosporin A (CsA). Inhibition of the AT1-receptor did not reduce AngII-mediated eNOS protein expression, whereas AT2 stimulation increased it 2.4-fold and AT2 inhibition suppressed it. The modulatory effects of the AT2-receptor on eNOS expression was confirmed in mice with a genetic deletion of the AT2-receptor (AT2-KO). In gel shift assays two putative NF-AT sites in a 1.6 kb eNOS promoter fragment showed NF-AT binding and a supershift by NF-AT2(-c1)-specific antibodies. Stimulation of transfected cells with AngII or specific AT2-receptor agonists resulted in a significant increase in eNOS promoter activity, which was blocked by CsA, MCIP1, and mutation of an upstream NF-AT site. CONCLUSION: 1) AngII-stimulation of the myocardium, both in vivo and in vitro, is accompanied by increased expression of eNOS. 2) This effect is mediated by the calcineurin pathway and is induced by the AT2-receptor. 3) These results define a calcineurin/NF-AT/eNOS pathway as downstream effector of AT2-receptor activation in the myocardium.


Assuntos
Calcineurina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Miocárdio/enzimologia , Óxido Nítrico Sintase/metabolismo , Proteínas Nucleares , Receptores de Angiotensina/metabolismo , Fatores de Transcrição/metabolismo , Angiotensina II/farmacologia , Animais , Animais Recém-Nascidos , Sequência de Bases , Sítios de Ligação/genética , Humanos , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Knockout , Miocárdio/citologia , Fatores de Transcrição NFATC , Óxido Nítrico Sintase/efeitos dos fármacos , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Regiões Promotoras Genéticas/genética , Ligação Proteica , Ratos , Receptor Tipo 2 de Angiotensina , Receptores de Angiotensina/genética , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Transfecção
17.
Mol Endocrinol ; 17(8): 1640-6, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12764077

RESUMO

In the adrenergic system, release of the neurotransmitter norepinephrine from sympathetic nerves is regulated by presynaptic inhibitory alpha2-adrenoceptors, but it is unknown whether release of epinephrine from the adrenal gland is controlled by a similar short feedback loop. Using gene-targeted mice we demonstrate that two distinct subtypes of alpha2-adrenoceptors control release of catecholamines from sympathetic nerves (alpha 2A) and from the adrenal medulla (alpha 2C). In isolated mouse chromaffin cells, alpha2-receptor activation inhibited the electrically stimulated increase in cell capacitance (a correlate of exocytosis), voltage-activated Ca2+ current, as well as secretion of epinephrine and norepinephrine. The inhibitory effects of alpha2-agonists on cell capacitance, voltage-activated Ca2+ currents, and on catecholamine secretion were completely abolished in chromaffin cells isolated from alpha 2C-receptor-deficient mice. In vivo, deletion of sympathetic or adrenal feedback control led to increased plasma and urine norepinephrine (alpha 2A-knockout) and epinephrine levels (alpha 2C-knockout), respectively. Loss of feedback inhibition was compensated by increased tyrosine hydroxylase activity, as detected by elevated tissue dihydroxyphenylalanine levels. Thus, receptor subtype diversity in the adrenergic system has emerged to selectively control sympathetic and adrenal catecholamine secretion via distinct alpha2-adrenoceptor subtypes. Short-loop feedback inhibition of epinephrine release from the adrenal gland may represent a novel therapeutic target for diseases that arise from enhanced adrenergic stimulation.


Assuntos
Glândulas Suprarrenais/metabolismo , Catecolaminas/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Sistema Nervoso Simpático/metabolismo , Agonistas de Receptores Adrenérgicos alfa 2 , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Tartarato de Brimonidina , Sinalização do Cálcio/fisiologia , Catecolaminas/sangue , Catecolaminas/urina , Células Cultivadas , Células Cromafins/efeitos dos fármacos , Células Cromafins/metabolismo , Epinefrina/sangue , Epinefrina/metabolismo , Epinefrina/urina , Exocitose/fisiologia , Retroalimentação Fisiológica , Camundongos , Camundongos Knockout , Norepinefrina/sangue , Norepinefrina/metabolismo , Norepinefrina/urina , Quinoxalinas/farmacologia , Receptores Adrenérgicos alfa 2/genética , Tirosina 3-Mono-Oxigenase/metabolismo
18.
Eur J Endocrinol ; 149(4): 343-50, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14514350

RESUMO

OBJECTIVE: Adrenaline inhibits insulin secretion through activation of alpha(2)-adrenoceptors (ARs). These receptors are linked to pertussis toxin-sensitive G proteins. Agonist binding leads to inhibition of adenylyl cyclase, inhibition of Ca(2+) channels and activation of K(+) channels. Recently, three distinct subtypes of alpha(2)-AR were described, alpha(2A)-AR, alpha(2B)-AR and alpha(2C)-AR. At present, it is unknown which of these alpha(2)-AR subtype(s) may regulate insulin secretion. We used mice deficient in alpha(2)-ARs to analyze the coupling and role of individual alpha(2)-AR subtypes in insulin-secreting beta cells. METHODS: The inhibitory effect of adrenaline on insulin secretion was measured in freshly isolated and cultured wild type (wt) and alpha(2)-AR knockout (KO) mouse islets in order to examine the receptor subtypes which mediate adrenaline-induced inhibition of insulin secretion. Adenylyl cyclase activity was measured in isolated cultured islets. Membrane potential was measured using the amphotericin B permeabilized patch clamp method in isolated and cultured single islet cells. RESULTS: In wt, alpha(2A)- and alpha(2C)-AR KO mouse islets, adrenaline, 1 microM/L, inhibited secretion by 83, 80 and 100% respectively. In contrast, in alpha(2A/2C)-AR double KO mouse islets, adrenaline had no effect on stimulated secretion indicating that both alpha(2A)-AR and alpha(2C)-AR, but not alpha(2B)-AR, are functionally expressed in mouse islets. Surprisingly, glucose (16.7 mM/L)-induced secretion in the presence of 1 microM/L forskolin was greatly impaired in alpha(2A)-AR KO islets. However, when cAMP levels were increased further by the combination of forskolin (5 microM/L) and 3-isobutyl-1-methylxanthine (100 microM/L), secretion was stimulated 2.7-fold (8.5-fold in wt islets). Adrenaline lowered the concentration of cAMP in wt and alpha(2C)-AR KO mouse islets by 74%. Adrenaline also hyperpolarized wt and alpha(2C)-AR KO beta cells. In contrast, adrenaline did not inhibit adenylyl cyclase in islets of alpha(2A)-AR KO mice, nor did it hyperpolarize alpha(2A)-AR KO beta cells. CONCLUSION: Adrenaline inhibits insulin release through alpha(2A)- and alpha(2C)-ARs via distinct intracellular signaling pathways.


Assuntos
Insulina/metabolismo , Receptores Adrenérgicos alfa 2/deficiência , Receptores Adrenérgicos alfa 2/fisiologia , Transdução de Sinais , 1-Metil-3-Isobutilxantina/farmacologia , Potenciais de Ação/efeitos dos fármacos , Adenilil Ciclases/metabolismo , Anfotericina B , Animais , Células Cultivadas , Colforsina/farmacologia , AMP Cíclico/metabolismo , Epinefrina/farmacologia , Glucose/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp
19.
Exp Neurol ; 247: 80-90, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23570902

RESUMO

Blood-brain-barrier (BBB) disruption, inflammation and thrombosis are important steps in the pathophysiology of acute ischemic stroke but are still inaccessible to therapeutic interventions. Rolipram specifically inhibits the enzyme phosphodiesterase (PDE) 4 thereby preventing the inactivation of the intracellular second messenger cyclic adenosine monophosphate (cAMP). Rolipram has been shown to relief inflammation and BBB damage in a variety of neurological disorders. We investigated the therapeutic potential of rolipram in a model of brain ischemia/reperfusion injury in mice. Treatment with 10mg/kg rolipram, but not 2 mg/kg rolipram, 2 h after 60 min of transient middle cerebral artery occlusion (tMCAO) reduced infarct volumes by 50% and significantly improved clinical scores on day 1 compared with vehicle-treated controls. Rolipram maintained BBB function upon stroke as indicated by preserved expression of the tight junction proteins occludin and claudin-5. Accordingly, the formation of vascular brain edema was strongly attenuated in mice receiving rolipram. Moreover, rolipram reduced the invasion of neutrophils as well as the expression of the proinflammatory cytokines IL-1ß and TNFα but increased the levels of TGFß-1. Finally, rolipram exerted antithrombotic effects upon stroke and fewer neurons in the rolipram group underwent apoptosis. Rolipram is a multifaceted antiinflammatory and antithrombotic compound that protects from ischemic neurodegeneration in clinically meaningful settings.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Lesões Encefálicas/prevenção & controle , Encefalite/prevenção & controle , Inibidores da Fosfodiesterase 4/uso terapêutico , Rolipram/uso terapêutico , Acidente Vascular Cerebral/patologia , Trombose/prevenção & controle , Animais , Edema Encefálico/etiologia , Edema Encefálico/prevenção & controle , Lesões Encefálicas/etiologia , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Encefalite/etiologia , Endotelina-1/genética , Endotelina-1/metabolismo , Hemodinâmica/efeitos dos fármacos , Infarto da Artéria Cerebral Média/complicações , Fluxometria por Laser-Doppler , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Ocludina/genética , Ocludina/metabolismo , Acidente Vascular Cerebral/complicações , Acidente Vascular Cerebral/etiologia , Trombose/etiologia
20.
J Cereb Blood Flow Metab ; 31(10): e1-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21792243

RESUMO

We assessed the neuroprotective potential of α(2)-adrenoceptors in ischemic stroke using mice with targeted deletions of individual α(2)-adrenoceptor subtypes (α(2A)(-/-), α(2B)(-/-), α(2C)(-/-), α(2A/C)(-/-)). The effects of the α(2)-adrenoceptor agonist clonidine were studied in parallel. Focal cerebral ischemia was induced with or without clonidine pretreatment by transient middle cerebral artery occlusion. Neurologic outcome and infarct volumes were evaluated on day 1. Cerebral blood flow (CBF) and mean arterial pressure were determined. α(2)-Adrenoceptor null mice did not display larger infarct volumes compared with wild-type (WT) mice under basal conditions (P>0.05). In line with this finding, pretreatment with clonidine did not protect from ischemic brain damage in WT mice or α(2A)(-/-), α(2B)(-/-), and α(2C)(-/-) mice. Clonidine induced smaller infarct volumes only in α(2A/C)(-/-) mice (P<0.05), but this did not translate into improved neurologic function (P>0.05). Importantly, while clonidine caused a significant decrease in arterial blood pressure in all groups, it had no blood pressure lowering effect in α(2A/C)(-/-) mice, and this correlated with higher CBF and smaller infarct volumes in this group. In summary, we could not demonstrate a neuroprotective function of α(2)-adrenoceptors in focal cerebral ischemia. Careful controlling of physiological parameters relevant for stroke outcome is recommended in experimental stroke studies.


Assuntos
Barreira Hematoencefálica/metabolismo , Isquemia Encefálica/metabolismo , Endotélio Vascular/metabolismo , Matriz Extracelular/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Barreira Hematoencefálica/patologia , Isquemia Encefálica/patologia , Adesão Celular , Claudina-5 , Endotélio Vascular/patologia , Matriz Extracelular/patologia , Integrina beta1/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Acidente Vascular Cerebral/patologia , Junções Íntimas/metabolismo , Junções Íntimas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA